Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
PLoS One ; 5(4): e10221, 2010 Apr 20.
Article in English | MEDLINE | ID: mdl-20421926

ABSTRACT

BACKGROUND: The transcription factor Snail1 induces epithelial-to-mesenchymal transition (EMT), a process responsible for the acquisition of invasiveness during tumorigenesis. Several transcriptomic studies have reported Snail1-regulated genes in different cell types, many of them involved in cell adhesion. However, only a few studies have used proteomics as a tool for the characterization of proteins mediating EMT. METHODOLOGY/PRINCIPAL FINDINGS: We identified by proteomic analysis using 2D-DIGE electrophoresis combined with MALDI-TOF-TOF and ESI-linear ion trap mass spectrometry a number of proteins with variable functions whose expression is modulated by Snail1 in SW480-ADH human colon cancer cells. Validation was performed by Western blot and immunofluorescence analyses. Snail1 repressed several members of the 14-3-3 family of phosphoserine/phosphothreonine binding proteins and also the expression of the Proliferation-associated protein 2G4 (PA2G4) that was mainly localized at the nuclear Cajal bodies. In contrast, the expression of two proteins involved in RNA processing, the Cleavage and polyadenylation specificity factor subunit 6 (CPSF6) and the Splicing factor proline/glutamine-rich (SFPQ), was higher in Snail1-expressing cells than in controls. The regulation of 14-3-3epsilon, 14-3-3tau, 14-3-3zeta and PA2G4 by Snail1 was reproduced in HT29 colon cancer cells. In addition, we found an inverse correlation between 14-3-3sigma and Snail1 expression in human colorectal tumors. CONCLUSIONS/SIGNIFICANCE: We have identified a set of novel Snail1 target proteins in colon cancer that expand the cellular processes affected by Snail1 and thus its relevance for cell function and phenotype.


Subject(s)
Colonic Neoplasms/chemistry , Gene Expression Regulation, Neoplastic , Neoplasm Proteins/analysis , Proteomics/methods , Transcription Factors/analysis , Electrophoresis, Gel, Two-Dimensional , Epithelial Cells , Humans , Mesenchymal Stem Cells , Repressor Proteins , Snail Family Transcription Factors , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Transcription Factors/physiology
2.
Anticancer Res ; 28(5A): 2613-23, 2008.
Article in English | MEDLINE | ID: mdl-19035286

ABSTRACT

Colorectal cancer is a major health problem worldwide. Aberrant activation of the Wingless-type mouse mammary tumour virus integration site family (Wnt)/beta-catenin signalling pathway due to mutation of adenomatous polyposis coli (APC), beta-catenin (CTNNB1) or AXIN genes is the most common and initial alteration in sporadic colorectal tumours. Numerous epidemiological and experimental studies have indicated a protective action of vitamin D against colorectal cancer. Previous work has demonstrated that the most active vitamin D metabolite, 1alpha,25-dihydroxyvitamin D3 (1,25(OH)2D3) inhibits beta-catenin transcriptional activity by promoting vitamin D receptor (VDR) binding to beta-catenin and the induction of E-cadherin expression. Recently, 1,25(OH)2D3 has been shown to distinctly regulate two genes encoding the extracellular Wnt inhibitors DICKKOPF-1 and DICKKOPF-4 (DKK-1, DKK-4). By an indirect transcriptional mechanism, 1,25(OH)2D3 increases the expression of DKK-1 RNA and protein, which acts as a tumour suppressor in human colon cancer cells harbouring endogenous mutations in the Wnt/beta-catenin pathway. In contrast, 1,25(OH)2D3 represses DKK-4 transcription by inducing direct VDR binding to its promoter. Unexpectedly, DKK-4 is a target of the Wnt/beta-catenin pathway and is up-regulated in colorectal tumours, and it has been shown to increase cell migration and invasion and to promote a proangiogenic phenotype. Together, these results show that 1,25(OH)2D3 exerts a complex set of regulatory actions leading to the inhibition of the Wnt/beta-catenin pathway in colon cancer cells that is in line with its protective effect against this neoplasia.


Subject(s)
Calcitriol/metabolism , Colonic Neoplasms/genetics , Gene Expression Regulation, Neoplastic , Intercellular Signaling Peptides and Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/metabolism , Colonic Neoplasms/metabolism , Humans , Intercellular Signaling Peptides and Proteins/biosynthesis , Signal Transduction
3.
Differentiation ; 75(3): 193-207, 2007 Mar.
Article in English | MEDLINE | ID: mdl-17288543

ABSTRACT

1alpha,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)), the most active vitamin D metabolite, regulates proliferation, survival, and differentiation in many cell types. 1,25(OH)(2)D(3) and several less calcemic analogs are in clinical trials against various neoplasias. We studied the effects of 1,25(OH)(2)D(3) on a panel of human breast cancer cells, which show similar vitamin D receptor (VDR) content but variable transcriptional and anti-proliferative responsiveness. In MDA-MB-453 cells, one of the responsive lines, 1,25(OH)(2)D(3) increased cell and nuclear size and induced a change from a rounded to a flattened morphology. By phase contrast, laser confocal and electron microscopy, we found that 1,25(OH)(2)D(3) changed the cytoarchitecture of actin filaments and microtubules and nuclear shape, induced filopodia and lamellipodia, and promoted cell-to-cell contacts via large cytoplasmic extensions. However, although claudin-7 and occludin content in the cells increased upon exposure to 1,25(OH)(2)D(3), these proteins were not located at the plasma membrane probably due to the absence of E-cadherin expression. Additionally, 1,25(OH)(2)D(3) induced the accumulation of alpha(v)-integrin, beta(5)-integrin, focal adhesion kinase (FAK), and paxillin in focal adhesion plaques, concomitant with the increased phosphorylation of the FAK. 1,25(OH)(2)D(3) enhanced MDA-MB-453 and MDA-MB-468 cell adhesion to plastic but decreased adhesion to laminin. The expression of the mesenchymal marker N-cadherin and of the myoepithelial marker P-cadherin was down-regulated by 1,25(OH)(2)D(3) in several breast cancer cell lines. Other myoepithelial proteins such as alpha(6)-integrin, beta(4)-integrin, and smooth muscle alpha-actin (SMA) were also repressed by 1,25(OH)(2)D(3) in MDA-MB-453 and MDA-MB-468 cells. Accordingly, mice lacking VDR (Vdr(-/-)) showed abnormally high levels of SMA and P-cadherin in their mammary gland. These findings show that 1,25(OH)(2)D(3) profoundly affects the phenotype of breast cancer cells, and suggest that it reverts the myoepithelial features associated with more aggressive forms and poor prognosis in human breast cancer.


Subject(s)
Breast Neoplasms/pathology , Calcitriol/pharmacology , Animals , Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Cadherins/antagonists & inhibitors , Cadherins/metabolism , Cell Movement , Cell Proliferation/drug effects , Cell Size , Female , Focal Adhesions/metabolism , Humans , Immunohistochemistry , Mice , Phenotype , Tumor Cells, Cultured , Vitamin D Response Element
4.
Front Biosci ; 10: 2723-49, 2005 Sep 01.
Article in English | MEDLINE | ID: mdl-15970529

ABSTRACT

1alpha,25-dihydroxyvitamin D3 (1,25(OH)2D3, Calcitriol) is a pleiotropic hormone with anti-proliferative, pro-apoptotic and pro-differentiation effects on numerous cell types, which suggest anti-cancer activity in addition to its classical regulatory action on calcium and phosphate metabolism. 1,25(OH)2D3 exerts its actions mainly via its high affinity receptor VDR through a complex network of genomic (transcriptional and post-transcriptional) and also non-genomic mechanisms, which are partially coincident in the different cells and tissues studied. Epidemiological and experimental in vitro and in vivo data support a cancer preventive role of 1,25(OH)2D3. The anti-cancer activity of 1,25(OH)2D3 and multiple analogs with reduced calcemic properties, which are thus less toxic, is under investigation in a long list of cultured cell types and in several in vivo models of wild-type and genetically-modified animals. Some vitamin D compounds have reached clinical trials, but results are still scarce.


Subject(s)
Antineoplastic Agents/pharmacology , Neoplasms/pathology , Vitamin D/analogs & derivatives , Animals , Antineoplastic Agents/therapeutic use , Clinical Trials as Topic , Disease Models, Animal , Humans , Neoplasm Transplantation , Neoplasms/drug therapy , Tumor Cells, Cultured , Vitamin D/pharmacology , Vitamin D/therapeutic use , Xenograft Model Antitumor Assays
5.
Oncogene ; 24(6): 1098-103, 2005 Feb 03.
Article in English | MEDLINE | ID: mdl-15592505

ABSTRACT

Wnt glycoproteins regulate homeostasis and development by binding to membrane Frizzled-LRP5/6 receptor complexes. Wnt signaling includes a canonical pathway involving cytosolic beta-catenin stabilization, nuclear translocation and gene regulation, acting as a co-activator of T-cell factor (TCF) proteins, and noncanonical pathways that activate Rho, Rac, JNK and PKC, or modulate Ca(2+) levels. DICKKOPF-1 (DKK-1) encodes a secreted Wnt antagonist that binds to LRP5/6 and induces its endocytosis, leading to inhibition of the canonical pathway. We show that activation of canonical signaling by Wnt1 or ectopic expression of active beta-catenin, TCF4 or LRP6 mutants induces transcription of the human DKK-1 gene. Multiple beta-catenin/TCF4 sites in the DKK-1 gene promoter contribute to this activation. In contrast, Wnt5a, which signals through noncanonical pathways, does not activate DKK-1. Northern and Western blot studies show that activation of the Wnt/beta-catenin pathway by treatment with lithium or Wnt3a-conditioned medium, or by stable expression of either Wnt1 or beta-catenin, increases DKK-1 RNA and protein, thus initiating a negative feedback loop. However, we found that DKK-1 expression decreases in human colon tumors, which suggests that DKK-1 acts as a tumor suppressor gene in this neoplasia. Our data indicate that the Wnt/beta-catenin pathway is downregulated by the induction of DKK-1 expression, a mechanism that is lost in colon cancer.


Subject(s)
Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Cytoskeletal Proteins/pharmacology , DNA-Binding Proteins/pharmacology , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Proteins/genetics , Trans-Activators/pharmacology , Transcription Factors/pharmacology , Down-Regulation , Genes, Tumor Suppressor , Helix-Loop-Helix Motifs , Homeostasis , Humans , Intercellular Signaling Peptides and Proteins/pharmacology , Signal Transduction , TCF Transcription Factors , Transcription Factor 7-Like 2 Protein , Wnt Proteins , Wnt1 Protein , beta Catenin
6.
J Biol Chem ; 279(16): 16638-45, 2004 Apr 16.
Article in English | MEDLINE | ID: mdl-14764591

ABSTRACT

Norwalk virus is a major cause of acute gastroenteritis for which effective treatments are sorely lacking. To provide a basis for the rational design of novel antiviral agents, the main replication enzyme in Norwalk virus, the virally encoded RNA-dependent RNA polymerase (RdRP), has been expressed in an enzymatically active form, and its structure has been crystallographically determined both in the presence and absence of divalent metal cations. Although the overall fold of the enzyme is similar to that seen previously in the RdRP from rabbit hemorrhagic disease virus, the carboxyl terminus, surprisingly, is located in the active site cleft in five independent copies of the protein in three distinct crystal forms. The location of this carboxyl-terminal segment appears to interfere with the binding of double-stranded RNA in the active site cleft and may play a role in the initiation of RNA synthesis or mediate interactions with accessory replication proteins.


Subject(s)
Norwalk virus/enzymology , RNA-Dependent RNA Polymerase/chemistry , Animals , Binding Sites , Escherichia coli , Models, Molecular , Protein Conformation , RNA-Dependent RNA Polymerase/metabolism , Rabbits , Substrate Specificity
SELECTION OF CITATIONS
SEARCH DETAIL
...